Você está na página 1de 9

See discussions, stats, and author profiles for this publication at: https://www.researchgate.

net/publication/7954388

Assays of matrix metalloproteinases (MMPs) activities: A review

Article  in  Biochimie · March 2005


DOI: 10.1016/j.biochi.2005.01.007 · Source: PubMed

CITATIONS READS

79 1,710

3 authors, including:

Joëlle Saulnier
Claude Bernard University Lyon 1
38 PUBLICATIONS   466 CITATIONS   

SEE PROFILE

All content following this page was uploaded by Joëlle Saulnier on 07 November 2017.

The user has requested enhancement of the downloaded file.


Biochimie 87 (2005) 265–272
www.elsevier.com/locate/biochi

Assays of matrix metalloproteinases (MMPs) activities: a review


Carine Lombard, Joëlle Saulnier, Jean Wallach *
Laboratoire de biochimie analytique et synthèse bioorganique, UFR Chimie-Biochimie, Université Claude-Bernard Lyon 1,
69622 Villeurbanne cedex, France

Received 26 October 2004; accepted 13 January 2005

Abstract

Measurement of matrix metalloproteinase (MMP) activity often remains a challenge, mainly in complex media. Two sets of methods are
currently used. The first one measures the hydrolysis of natural protein substrates (labeled or not) and includes the popular zymography. These
techniques which are quite sensitive, cannot generally be carried out on a continuous basis. The second one takes mainly advantage of the
increase of fluorescence, which is associated to the hydrolysis of initially quenched fluorogenic peptide substrates. Quite recently, another
group, which is a compromise between the other two, has been developed. It measures the hydrolysis of synthetic triple-helical peptide
substrates. These different methods are described and discussed.
© 2005 Elsevier SAS. All rights reserved.

Keywords: Metalloprotease assay; Fluorescent substrates; Immunocapture; Triple-helical peptides

1. Introduction not always figuring the biological activity of the enzymes,


and more cumbersome and complex methods using native or
The choice of an assay for measuring the activity of an modified proteins which are more difficult to use for screen-
enzyme is critically depending on the qualitative and quanti- ing. In the case of studies aimed at determining the role of
tative objectives of the study. This is particularly important in MMPs, it is obviously better to use native proteins or deriva-
the case of the matrix metalloproteinases (MMPs), as their tives as substrates whereas the screening of inhibitors, in pres-
natural substrates are usually insoluble (or poorly soluble) ence of purified enzymes, will be easier with synthetic sub-
proteins, complex mixtures of proteins and associate macro- strates, which allow to perform rapid and numerous assays
molecules, structural components of extracellular matrices. within quite a short time and high-throughput screening. The
They are synthesized as inactive zymogens and must be enzy- situation becomes more complicated when assays are carried
matically activated for being active. Furthermore, their activi- out in biological fluids, due to the presence of interfering mol-
ties are in vivo regulated by endogenous inhibitors (TIMPs). ecules, mainly inhibitors and also due to the fact that MMP
For measuring their enzymatic activity, the choice is between specificities are similar.
easy-to-use techniques, generally with synthetic substrates, In many circumstances, immunochemical procedures can
replace enzymatic assays, but they cannot discriminate
between zymogens and active enzymes.
Abbreviations: A2pr, 2,3-diaminopropionic acid; Adp, 2-amino-3-(6,7- The different assays of matrix metalloproteinase activities
dimethoxy-4-coumaryl)propionic acid; APMA, p-aminophenylmercuric ace-
may be roughly classified in:
tate; Ctmr, 5-carboxytetramethylrhodamine; DNFB, dinitrofluorobenzene;
Dnp, dinitrophenyl; Dpa, diphenylalanine; DTNB, dithiobis-(2-nitrobenzoic • Assays using proteins (modified or not);
acid); FIA, flow-injection analysis; FITC, fluorescein isothiocyanate; HTS, • Assays using synthetic linear peptides and their deriva-
high-throughput screening; HRP, horseradish peroxidase; Mca, 7-methoxy- tives;
coumarin-2-acetic acid; MOCAc, (7-methoxycoumarin-4-yl)acetyl; Nma, • Assays using synthetic “mini-collagens”.
N-methylanthranilic acid; TNBSA, 2,4,6-trinitrobenzene sulfuric acid; TNF,
tumor necrosis factor; uPA, urokinase-type plasminogen activator.
In this brief review, we will describe, illustrate, and compare
* Corresponding author. these assays, referring to recent published results from vari-
E-mail address: jean.wallach@univ-lyon1.fr (J. Wallach). ous laboratories.
0300-9084/$ - see front matter © 2005 Elsevier SAS. All rights reserved.
doi:10.1016/j.biochi.2005.01.007
266 C. Lombard et al. / Biochimie 87 (2005) 265–272

2. Assays using proteins detection limit was lowered to 2 pg. Initial rates of hydrolysis
could be determined from density measurements within 30 h
Ideally, it would be better to measure the MMP activities of digestion for enzyme concentrations ranging from 0.1 to
with their most specific native proteins as substrates. But the 4 ng MMP-2 [8]. In a similar way, but using a single-step
hydrolysis rates are low and the classical methods are not staining method, which led to fast and reproducible results,
sensitive enough. This is the reason why current methods pref- Leber and Balkwill [9] could demonstrate a detection limit
erably use labeled proteins, whereas some other original pro- of 32 pg for pro-MMP-9 and a linear range below 1 ng. More
cedures have also been described. recently, Gogly et al. reported a detection limit as low as 0.1 pg
in the case of an MMP-1 assay on collagen zymograms. Such
2.1. Non-modified proteins a sensitivity is comparable to an immunodot blot assay using
chemiluminescence but it may be noticed that immunoas-
While collagenolytic activity may be evaluated by the spec- says do not discriminate between pro- and active forms of
trophotometric assay of hydroxyproline in the hydrolysates, enzyme which is not the case for zymography, in the experi-
this technique is more valuable for bacterial metalloprotein- mental conditions described by the authors. The specificity
ases than for matrix ones [1]. Contrastingly, cleavage rates of of this method for collagenase was demonstrated, as no lysis
insoluble and soluble collagens by tissue collagenases could could be observed with 300 pg of either MMP-2 or MMP-3
be measured from dansylation at the N-terminal residues of [10].
the cleaved peptides, after hydrolysis. Extensive hydrolysis Unlike gelatinases, matrilysin (MMP-7), and collagena-
and HPLC separation were monitored by fluorescence mea- ses (MMP-1 and MMP-13) are difficult to detect. In a recent
surements. Using such a technique, kinetic parameters could report, it was demonstrated that heparin may enhance the
be determined (KM values were calculated to be in the micro- assays, lowering the detection limit to 30 pg for MMP-7 and
molar range) [2]. A similar approach has been conducted by to 0.2 ng for MMP-1 and MMP-13. The mechanism of activ-
Mecham et al., in order to characterize the cleavage sites in ity enhancement is not fully explained, but it has been sug-
insoluble elastin. Prior to Edman sequencing of hydrolyzed gested that heparin may act by inducing a conformation
peptides, they used DNFB, which reacts covalently with pri- change, facilitating refolding, reducing autolysis or helping
mary and secondary amino groups. They could then identify anchoring of the enzyme to the gel [11].
′ Zymography has also been applied to in situ studies. Ini-
the P1 residues of proteolytic hydrolysis. Theoretically, it
would be possible to quantify the cleavage rates, even if it tially, limited to soft and homogenous tissues, it has recently
would obviously be a tedious method [3]. been used to demonstrate the sub-lamellar location of gelati-
Very recently, three collagenase assays (MMP-1, MMP-8, nases in hard epidermal layers and a comparison has been
and MMP-13) with triple-helical collagens as substrates, have made with an adjacent soft connective tissue matrix. The
been developed using capillary gel electrophoresis with laser- authors could then evaluate the influence of tissue topogra-
induced dynamic fluorescence detection, with a non-covalent phy on the technique [12].
fluorescent dye (NanoOrange). A good linearity of peak areas Zymography may also be performed in a reverse mode,
was obtained over each assay range (15–150 ng/tube for designed to detect the presence of metalloproteinase inhibi-
MMP-1, 3-30 ng/tube for MMP-8, and 1.5–30 ng/tube for tors. In this case, a proteinase is directly incorporated into the
MMP-13). The method has been applied to the estimation of substrate-containing gel. The method was applied for the
inhibitors and the results well agree with those obtained with quantitative assessment of metalloproteinase inhibitor activ-
fluorescent peptides [4]. ity either with gelatin [13] or FITC-labeled collagen and
A great number of data concerning MMP activities has casein [14] as substrates.
been obtained by zymography, which allows the visualiza- Today, zymography is probably one of the most popular
tion of protease activities. This technique involves the elec- techniques for measuring MMP activities. Some experimen-
trophoresis of enzymes through polyacrylamide gels contain- tal procedures have recently been detailed and discussed in
ing copolymerized protease substrate. It has been used for the case of enzymatic activities in a crude medium of cul-
many years mainly with gelatin as a substrate. Electrophore- tured synoviocytes [15].
sis is generally performed in sodium dodecylsulfate (SDS) 2.2. Labeled proteins
and proteases are then renatured, enabling the hydrolysis of
the substrate. After staining, the zones of digestion may be Various methods have been developed, using chemically
evidenced and/or quantified. In the early 90s, zymography modified or engineered proteins. In the first category, the most
was used to analyze the gelatinolytic activities of secreted popular are proteins labeled either by radioelements or by
proteinases of metastatic cell lines [5–7], but it was only in fluorescent groups, but recent results have emphasized the
1994 that the technique became quantitative. With the help of usefulness of other modifications.
a purified MMP-2/TIMP-2 complex, Kleiner and Stetler-
Stevenson could detect picogram quantities of the enzyme. A 2.2.1. Radiolabeled proteins
linear relationship could be evidenced between 10 and 120 pg For sensitivity and ease of detection, labeling by means of
after a 18 h-digestion period. After a 43 h-incubation, the [3H] acetic anhydride is frequently performed. Assays have
C. Lombard et al. / Biochimie 87 (2005) 265–272 267

been developed with casein [16], collagens (native or dena- TNBSA, due to the liberation of new reactive amino groups.
tured) [17] and elastin [3]. It is also possible to use [125I] Applied to MMP-2 and -9, the assay is quite rapid (<1 h),
gelatin [18]. In any case, undegraded proteins are precipi- specific and may be used for the evaluation of inhibition [25].
tated with trichloracetic acid and radioactivity counted in the This methodology has recently been applied to measure the
supernatant. inhibition of MMP-1, -2 and -9 by means of pycnogenol
metabolites [26].
2.2.2. Fluorescently labeled proteins
Fluorescein and other derivatives have been used for a long 2.2.5. Immunocapture assay
time as protein modifiers, allowing sensitive fluorometric A new general principle for measurement of proteolytic
assays. Nevertheless, all these assays require a precipitation activities was developed in 1997 by Verheijen et al. [27]. Sum-
step, keeping them as endpoint assays. For a continuous moni- marized in Fig. 1, it is based on the activation of pro-
urokinase in urokinase and further spectrophotometric assay,
toring of the reaction, it has recently been proposed to use
using a specific chromogenic peptide substrate. The method
dyes, which are linked in such a way that fluorescence is
may be extended to metalloproteinase assay by replacing the
quenched. Protease-catalyzed hydrolysis relieves this quench-
activation site present in pro-urokinase-type plasminogen acti-
ing, yielding a dramatic increase in fluorescence [19]. Deriva-
vator (pro-uPA), namely Pro-Arg-Phe-Lys-Ile-Ile-Gly-Gly, by
tives of elastin, collagen, and gelatin are now commercially
a sequence recognized by MMPs. A comparison between pri-
available (Molecular Probes and others) and enzymatic activ-
mary sequences of the recognition site of various MMPs led
ity may be monitored with fluorescence microplate readers
the authors to choose the introduction of the sequence: Arg-
or spectrofluorometers.
Pro-Leu-Gly-Ile-Ile-Gly-Gly in a mutant pro-uPA, obtained
Flow cytometric analysis is another method for measuring
with the recombinant circle PCR method. A coupled assay
the MMP activity with fluorescent substrates, such as fluo-
was then developed with pyroGlu-Gly-Arg-pNA as a sub-
rescein isothiocyanate (FITC)-labeled gelatin, after coating
strate for modified urokinase. The parabolic curve absor-
on polystyrene microspheres [20]. This technique has recently
bance vs. time could be transformed into a linear assay by
been employed as a routine method to measure MMP-2 and
plotting absorbance changes against square time. The sensi-
MMP-9 activities and MMP-9 inhibition by means of a pep-
tivity was the highest for MMP-2 and MMP-9, intermediate
tidomimetic based on the consensus sequence of the cleav-
for MMP-1 and much lower for MMP-3 and MMP-7. The
age sites in type II collagen [21].
assay could be made specific for MMP-9 [28] and MMP-2
[29] using specific monoclonal antibodies. By means of these
2.2.3. Biotinylated gelatin
antibodies, MMPs were captured from complex media and
A simple, inexpensive, and highly sensitive assay of
activities specifically analysed. Simple modifications could
MMP-2 (and, at a lower extent, of MMP-9), using biotiny-
raise the sensitivity to 60 pg ml–1 for MMP-9 activity assay
lated gelatin, was reported in 2000 by Ratnikov et al. [22].
[30]. This method, now commercially available (Amersham
After a proteolytic hydrolysis, steptavidin-coated plates were
BioSciences) has been further successfully used for activity
used to capture the hydrolyzed biotin-containing fragments.
measurements and inhibitor assessments [31,32].
Activity was measured with a streptavidin-peroxidase (HRP)
complex added to the medium. As two biotin residues are
necessary for binding both streptavidin and streptavidin- 3. Assays using linear peptides
HRP, the hydrolysis leads to cleavage products devoid of
biotin or possessing only one biotin and a decrease in absor- The first substrates used for measuring peptidasic MMPs
bance is observed. This 2 h-assay allowed a measurement of activities were linear peptides corresponding to the scissible
the gelatinolytic MMP-2 activity for concentrations as low as
0.16 ng ml–1. Recently, with minor modifications, this tech-
nique was applied to the demonstration of the activation of
pro-MMP-9 by APMA or trypsin. The authors could obtain a
linear relationship of the absorbance decrease vs. the enzyme
concentration within the range 0.15–2.5 ng ml–1 [23].

2.2.4. Succinylated gelatin


This chemical modification has been proposed in order to
develop a spectrophotometric assay, based on the measure-
ment of primary amines exposed after gelatinase hydrolysis.
Previously, applied to elastin hydrolysis by elastase [24], sub-
strate succinylation is performed with succinic anhydride,
blocking free amino groups, which cannot then react with
TNBSA. When succinylated gelatin is enzymatically hydro- Fig. 1. Colorimetric assay of matrix metalloproteinase activity using modi-
lyzed, absorbance at 450 nm increases after the action of fied pro-urokinase.
268 C. Lombard et al. / Biochimie 87 (2005) 265–272

Gly-Ile region of collagen and N-terminally labeled by a Dnp synthesized, EDANS being introduced either on the side chain
group, such as Dnp-Pro-Gln-Gly-Ile-Ala-Gly-Gln-D-Arg. In of Glu or at the C-terminal end of the peptides [45–47].
this case, the Dnp-Pro-Gln-Gly peptide was either extracted Two other couples have been introduced: Lucifer
into ethylacetate and then spectrophotometrically assayed at Yellow/Ctmr and more recently two CyDye fluors. In the first
365 nm [33] or separated by HPLC from the reaction mixture case, the enzyme-specific peptide sequence is placed between
and estimated from its peak area after calibration [34]. Other a N-terminal Ser and C-terminal Lys. Serine is further oxi-
similar peptides were suggested including the Gly-Ile bond, dized by periodate and coupled with Lucifer Yellow, forming
such as Dnp-Pro-Leu-Gly-Ile-Ala-Gly for MMP-8 and Dnp- a hydrazone. C-terminal Lys is then coupled with 5-carboxy-
Pro-Gln-Gly-Ile-Ala for MMP-9 [35]. Weingarten et al. pro- tetramethylrhodamine, quenching the dye fluorescence
posed a series of peptide derivatives, including thiopeptolide, [48,49]. Despite its interesting properties, this couple has not
which has a thioester bond instead of the cleavage site pep- been extensively used for MMP assays.
tide bond. After hydrolysis, the liberated thiol reacts with 5,5′- More recently, CyDye fluors were investigated for the study
dithiobis-(2-nitrobenzoic acid) (DTNB), forming a colored of enzyme activity and the development of high-throughput
2-nitro-5-thiobenzoic acid, which can be detected at 412 nm screening (HTS). The pair Cy3/Cy5Q was validated for MMP
[36,37]. A colorimetric assay kit based on this reaction is com- activity assays and a comparison made with the Mca/Dnp
mercially available for MMP-3 assay and kits for MMP-1, pair [50]. The system could be applied to a high-throughput
MMP-7 and MMP-12 were developed (BioMol). screening of MMP-3 inhibitors, with an imaging platform,
Nevertheless, most of MMPs assays using peptide sub- which gave improved reading accuracy and faster plate pro-
strates are fluorometric and based on resonance energy trans- cessing times compared with a commercially available
fer quenching. Such an effect occurs between a fluorescent microtitre plate reader [51].
donor group and a quenching acceptor, with an optimal dis- A recent review summarized the state of the art on the fluo-
tance between them. The hydrolysis of the substrate and the rogenic substrates, which has yet been described [52].
separation of both fragments abolish the energy transfer and Together with data on kinetic parameters, this paper is full of
lead to an increase of fluorescence. This general principle has practical information concerning the synthesis of substrates,
been applied for fluorimetric assays of many proteolytic their characterization and experimental procedures for assays.
enzymes [38]. Fluorometric assays are very convenient for MMP assays
Six different fluorophores have been used in MMP fluo- and for screening inhibitors. Nevertheless, it has been noticed
rogenic peptide substrates, together with various quenchers. that fluorescence changes are not linear with substrate con-
The first was tryptophan (Trp) with Dnp as the quenching centration, due to “inner filter effects”. It has then been sug-
group. Stack and Gray [39] demonstrated that the presence gested that apparent substrate inhibition observed by several
of Trp in P’2 position and of Dnp in P3 position leads to an research groups should be reconsidered. This phenomenon
efficient substrate for both continuous collagenase and gelati- has been described in particular with EDANS/DABCYL in
nase assays, with kcat/KM values of 5.4 and 440 µM–1 h–1, the case of cytomegalovirus protease [53] and trypsin [54]
respectively. The optimization of the peptide sequence allowed assays. Such an effect may influence the determination of
Netzel-Arnett et al. [40] to develop fluorescent assays for five Michaelis–Menten parameters, if the calibration curves of
MMPs. Dnp has also been associated with two other fluoro- fluorescence changes are not made vs. substrate concentra-
phores, namely (7-methoxycoumarin-4-yl)acetyl (Mca) [41] tion. The analysis of this effect on the activity vs. concentra-
and N-methylanthranilic acid (Nma) [42], these two groups tion was done by Geoghegan [49] in the case of the TNF
being much more fluorescent than tryptophan. In particular, convertase.
the substrate Mca-Pro-Leu-Gly-Leu-Dpa-Ala-Arg-NH2, was Although generally developed in cuvettes or microplates,
extensively used for monitoring MMP activities and for MMP activity may also be measured with fluorescent sub-
screening inhibitors. In 2004, Neumann et al. [43] improved strate peptides by flow injection analysis (FIA). Itoh et al.
the properties of this substrate by N-terminal addition of a [55] have proposed a simple and convenient method for mea-
Lys, increasing both the specificity constant and water solu- suring MMP-7 activity. Using MOCAc-Pro-Leu-Gly-Leu-
bility. A2pr (DNP)-Ala-Arg-NH2 as a substrate, fluorescence was
Another very popular donor/acceptor pair is EDANS: measured after a 2 min-hydrolysis, leading to a linear stan-
5-[(2-aminoethyl)amino] naphthalene-1-sulfonic acid and dard curve from 5 to 100 pmol of enzyme.
DABCYL: 4-(4-dimethylaminophenylazo)benzoic acid) (a
sulfonyl derivative (DABSYL) may also be used as a
quencher). This couple has many advantages: (1) there is a 4. Assays using triple-helical peptides
good spectral overlap between the EDANS emission spec-
trum and the DABCYL absorption spectrum, (2) EDANS has As mentioned earlier in this review, the study of the pre-
a high fluorescence, (3) the high wavelengths limit interfer- cise mechanism of action of MMPs and the evaluation of
ence with amino acid side chain fluorescence. First intro- inhibitors need the use of molecules which correspond, as
duced in a substrate for assaying HIV-1 protease [44], MMP closely as possible, to the native substrates of the enzymes. It
fluorogenic substrates including these groups have also been is the reason why well-defined triple helical peptides, derived
C. Lombard et al. / Biochimie 87 (2005) 265–272 269

Fig. 2. Schematic structure of branched triple-helical peptides.

from collagen sequences, were designed, mainly by Greg derivative as fluorophore. 2-Amino-3-(6,7-dimethoxy-4-
Fields and his group. The challenge was to synthesize struc- coumaryl)propionic acid (Adp), associated with Dnp as a
tures, which would be stable enough to remain in triple- quencher, definitely assumed to be compatible with multi-
helical conformation in the conditions of assays. well plate readers and better tolerated by MMPs than
Different approaches have been described. A solid-phase Lys(Mca) [70].
synthesis was first developed, featuring a C-terminal cova- Even if these substrates are not commercially available and
lent branch and incorporating specific sequences between this difficult to synthesize, they are essential to model the MMP
branch and a N-terminal (Gly-Pro-Hyp)n domain (Fig. 2) activity. Very recently, the importance of their thermal stabil-
[56,57]. ity on the activities of different MMP has been emphasized
Another approach was the design of “peptide-amphiphile” and may help demonstrate subtle differences in their abilities
triple helices, based on the non-covalent self-assembly of lipo- to hydrolyze triple helices [71]. Furthermore, the study indi-
philic molecules, N-terminally-linked to a peptide sequence cating that collagenase unwinds triple-helical collagen prior
[58–60]. Both the covalent branch and “peptide-amphiphile” to peptide bond hydrolysis, clearly supports the usefulness of
approaches have been used for studying susceptibility of triple-helical peptides as MMPs substrates to understand the
triple-helical peptides to MMP hydrolysis and localization of mechanism of action of MMPs [72].
enzyme cleavage sites, with Edman degradation sequence
analysis and MALDI-MS [61,62].
In order to develop a continuous assay method, Lauer-
5. Concluding remarks
Fields et al. [63] designed and synthesized triple-helical pep-
tides containing fluorophores. Fluorescence quenching used
One of the challenges of the MMP assays is the develop-
the Mca/Dnp pair, Lys(Dnp) and Lys(Mca) introduced in the
ment of sensitive, selective, and rapid methods.
positions P′5 and P5, respectively.
Another type of triple-helical structure was designed, based If we consider the above-mentioned methodologies, none
on a disulfide bridging approach. It contains a collagenase of them completely fulfil these requirements. The use of natu-
cleavage site linked to a cystine-knot and is N-terminally ral substrates mimics the true MMP activities but is generally
extended by (Gly-Pro-Hyp)5 triplets [64]. The addition of a tedious and slow. The use of fluorescent substrates is sensi-
Gly-Pro-Hyp repeat near the cystine knot leads to a remark- tive, rapid but does not always figure the in vivo activity of
ably stable triple-helical structure, with a melting tempera- the enzymes. The use of triple-helical peptides seems to be a
ture of 41 °C [65]. As previously, done by Fields and his team, good compromise between them, but these substrates are not
these substrates were made fluorogenic by addition of a dan- easy to synthesize and, as far as we know, not commercially
syl group and tryptophan, on the N-terminal position of the available yet.
a1 and a′1 chain, respectively [66]. In any case, these triple- Observations have been done concerning the role, in the
helical peptide substrates were hydrolyzed in a similar man- interactions proteins-MMPs, of residues distant from the pri-
ner to native collagens, cleavage occurring between Gly and mary recognition site. These relevant interactions are not
Ile (or Leu) according to the chain type. The kinetic param- present when synthetic substrates are used [73]. Moreover,
eters for hydrolysing several of them by various MMPs have when using fluorogenic substrates, Beekman et al. [74] could
recently been reviewed [67,68]. In the last 2 years, different measure MMP-13 activity, even when MMP-13 was in com-
improvements were made. As far as selectivity is concerned, plex with a2-macroglobulin. Such an activity could not be
two triple-helical peptides were designed as MMP-2 and evidenced with collagen as a substrate.
MMP-9 selective substrates to evaluate the active MMP pro- Direct comparison of fluorometric and zymographic meth-
duction in cellular systems [69]. ods led Quesada et al. [75] to conclude that gelatin zymogra-
At the same time, studies were carried out to limit the phy is more sensitive than fluorometric methods using either
increase of hydrophobicity due to the introduction of the Mca FITC-casein or fluorogenic peptides, but insisted on prob-
270 C. Lombard et al. / Biochimie 87 (2005) 265–272

lems associated with quantitation and they recommended the [11] W.H. Yu, J.F. Woessner Jr., Heparin-enhanced zymographic detection
use of fluorogenic substrates for high throughput screening of matrilysin and collagenases, Anal. Biochem. 293 (2001) 38–42.
[12] B.A. Mungall, C.C. Pollitt, In situ zymography: topographical consid-
of MMP inhibition. erations, J. Biochem. Biophys. Methods 47 (2001) 169–176.
A recent paper is suggesting another possible orientation [13] G.W. Oliver, J.D. Leferson, W.G. Stetler-Stevenson, D.E. Kleiner,
for improving MMP assays, particularly in complex mix- Quantitative reverse zymography: analysis of picogram amounts of
tures. Rasmussen et al. developed a new technique for mea- metalloproteinase inhibitors using gelatinase A and B reverse zymo-
grams, Anal. Biochem. 244 (1997) 161–166.
suring activity of a mixture of MMPs. Named MEMRAS
[14] S. Hattori, H. Fujisaki, T. Kiriyama, T. Yokoyama, S. Irie, Real-time
(“Multiple-Enzyme/Multiple-Reagent Assay System”), this zymography and reverse zymography: a method for detecting activi-
technique uses substrates with different selectivity profiles ties of matrix metalloproteinases and their inhibitors using FITC-
against a group of MMPs. If they solve simultaneous equa- labeled collagen and casein as substrates, Anal. Biochem. 301 (2002)
tions, the authors could estimate individual activities in a 27–34.
[15] L. Troeberg, H. Nagase, Measurement of matrix metalloproteinase
MMP mixture from independent initial rate experiments [76].
activities in the medium of cultured synoviocytes using zymography,
Another promising axis is the future development of Methods Mol. Biol. 225 (2003) 77–87.
protease-specific protein chips and protease-activity chips, as [16] D.H. Manicourt, V. Lefebvre, An assay for matrix metalloproteinases
suggested by Lopez-Otin and Overall [77]. With the help of and other proteases acting on proteoglycans, casein, or gelatin, Anal.
combinatorial peptide libraries, the improvement of sub- Biochem. 215 (1993) 171–179.
[17] J.B. Catterall, T.E. Cawston, Assays of matrix metalloproteinases
strate selectivity will favour such an approach. (MMPs) and MMP inhibitors: bioassays and immunoassays appli-
cable to cell culture medium, serum and synovial fluid, Methods Mol.
Biol. 225 (2003) 353–364.
Acknowledgments [18] T. Sorsa, T. Salo, E. Koivunen, J. Tyynelä, Y.T. Konttinen, U. Berg-
mann, et al., Activation of type IV procollagenases by human tumor-
We thank Mrs C. Beaufrere for English improvement. associated trypsin-2, J. Biol. Chem. 272 (1997) 21067–21074.
[19] D.A. Menges, D.L. Ternullo, A.L. Tan-Wilson, S. Gal, Continuous
assay of proteases using a microtiter plate fluorescence reader, Anal.
Biochem. 254 (1997) 144–147.
References [20] Y. St-Pierre, M. Desrosiers, P. Tremblay, P.-O. Esteve, G. Opdenakker,
Flow cytometric analysis of gelatinase B (MMP-9) activity using
[1] J.L. Hao, T. Nagano, M. Nakamura, N. Kumagai, H. Mishima, immobilized fluorescent substrate on microspheres, Cytometry 25
T. Nishida, Effect of galardin on collagen degradation by Pseudomo- (1996) 374–380.
nas aeruginosa, Exp. Eye Res. 69 (1999) 595–601. [21] J. Hu, P.E. Van den Steen, M. Houde, T.T. Ilenchuk, G. Opdenakker,
[2] S. Takahashi, S. Han, The rates of cleavage of insoluble and soluble Inhibitors of gelatinase B/matrix metalloproteinase-9 activity. Com-
collagens by tissue collagenases, Connect. Tissue Res. 30 (1993) parison of a peptidomimetic and polyhistidine with single-chain
99–116. derivatives of a neutralizing monoclonal antibody, Biochem. Pharma-
[3] R.P. Mecham, T.J. Broekelmann, C.J. Fliszar, S.D. Shapiro, H.G. Wel- col. 67 (2004) 1001–1009.
gus, R.M. Senior, Elastin degradation by matrix metalloproteinases. [22] B. Ratnikov, E. Deryugina, J. Leng, G. Marchenko, D. Dembrow,
Cleavage site specificity and mechanism of elastolysis, J. Biol. Chem. A. Strongin, Determination of matrix metalloproteinase activity using
272 (1997) 18071–18076. biotinylated gelatin, Anal. Biochem. 286 (2000) 149–155.
[4] M. Sano, I. Nishino, K. Ueno, H. Kamimori, Assay of collagenase [23] H.A. Bawadi, T.M. Antunes, F. Shih, J.N. Losso, In vitro inhibition of
activity for native triple-helical collagen using capillary gel electro- the activation of pro-matrix metalloproteinase 1 (Pro-MMP-1) and
phoresis with laser-induced fluorescence detection, J. Chromatogr. B pro-matrix metalloproteinase 9 (pro-MMP-9) by rice and soybean
809 (2004) 251–256. Bowman-Birk inhibitors, J. Agric. Food Chem. 52 (2004) 4730–4736.
[5] F. Umenishi, H. Yasumitsu, Y. Ashida, J. Yamauti, M. Umeda, [24] S.K. Rao, M. Mathrubutham, A. Karteron, K. Sorensen, J.R. Cohen, A
K. Miyazaki, Purification and properties of extracellular matrix- versatile microassay for elastase using succinylated elastin, Anal.
degrading metalloproteinase overproduced by Rous sarcoma virus- Biochem. 250 (1997) 222–227.
transformed rat liver cell line, and its identification as transin, J. [25] V.M. Baragi, B.J. Shaw, R.R. Renkiewicz, P.J. Kuipers, H.G. Welgus,
Biochem. (Tokyo) 108 (1990) 537–543. M. Mathrubutham, et al., A versatile assay for gelatinases using
[6] K. Miyazaki, Y. Hattori, F. Umenishi, H. Yasumitsu, M. Umeda, succinylated gelatin, Matrix Biol. 19 (2000) 267–273.
Purification and characterization of extracellular matrix-degrading [26] T. Grimm, A. Schäfer, P. Högger, Antioxidant activity and inhibition
metalloproteinase, matrin (pump-1), secreted from human rectal car- of matrix metalloproteinases by metabolites of maritime pine bark
cinoma cell line, Cancer Res. 50 (1990) 7758–7764. extract (pycnogenol), Free Radic. Biol. Med. 36 (2004) 811–822.
[7] Y. Kato,Y. Nakayama, M. Umeda, K. Miyazaki, Induction of 103-kDa [27] J.H. Verheijen, N.M.E. Nieuwenbroek, B. Beekman, R. Hanemaaijer,
gelatinase/type IV collagenase by acidic culture conditions in mouse H.W. Verspaget, H.K. Ronday, et al., Modified proenzymes as artifi-
metastatic melanoma cell lines, J. Biol. Chem. 267 (1992) 11424– cial substrates for proteolytic enzymes: colorimetric assay of bacterial
11430. collagenase and matrix metalloproteinase activity using modified
[8] D.E. Kleiner, W.G. Stetler-Stevenson, Quantitative zymography: pro-urokinase, Biochem. J. 323 (1997) 603–609.
detection of picogram quantities of gelatinases, Anal. Biochem. 218 [28] R. Hanemaaijer, H. Visser,Y.T. Konttinen, P. Koolwijk, J.H. Verheijen,
(1994) 325–329. A novel and simple immunocapture assay for determination of gelati-
[9] T.M. Leber, F.R. Balkwill, Zymography: a single-step staining nase B (MMP-9) activities in biological fluids: saliva from patients
method for quantitation of proteolytic activity on substrate gels, Anal. with Sjögren’s syndrome contain increased latent and active
Biochem. 249 (1997) 24–28. gelatinase-B levels, Matrix Biol. 17 (1998) 657–665.
[10] B. Gogly, N. Groult, W. Hornebeck, G. Godeau, B. Pellat, Collagen [29] S.J. Capper, J. Verheijen, L. Smith, M. Sully, H. Visser, R. Hane-
zymography as a sensitive and specific technique for the determina- maaijer, Determination of gelatinase A (MMP-2) activity using a
tion of subpicogram levels of interstitial collagenase, Anal. Biochem. novel immunocapture assay, Ann. N. Y. Acad. Sci. 878 (1999) 487–
255 (1998) 211–216. 490.
C. Lombard et al. / Biochimie 87 (2005) 265–272 271

[30] R. Hanemaaijer, N. Van Lent, H. Visser, J. Verheijen, Methods to [49] K.F. Geoghegan, Improved method for converting an unmodified
increase the sensitivity of the MMP-9 activity assay, Life Science peptide to an energy-transfer substrate for a proteinase, Bioconjug.
News 3 (1999) 8–9. Chem. 7 (1996) 385–391.
[31] M. Crul, L.V. Beerepoot, E. Stokvis, J.S.P. Vermaat, H. Rosing, [50] J. Peppard, Q. Pham, A. Clark, D. Farley, Y. Sakane, R. Graves, et al.,
J.H. Beijnen, et al., Clinical pharmacokinetics, pharmacodynamics Development of an assay suitable for high-throughput screening to
and metabolism of the novel matrix metalloproteinase inhibitor ABT- measure matrix metalloprotease activity, Assay Drug Dev. Technol. 1
518, Cancer Chemother. Pharmacol. 50 (2002) 473–478. (2003) 425–433.
[32] M.A. Huisman, A. Timmer, M. Zeinstra, E.K. Serlier, R. Hanemaaijer, [51] J. George, M.L. Teear, C.G. Norey, D.D. Burns, Evaluation of an
H. Van Goor, et al., Matrix-metalloproteinase activity in first trimester imaging platform during the development of a FRET protease assay, J.
placental bed biopsies in further complicated and uncomplicated Biomol. Screen. 8 (2003) 72–80.
pregnancies, Placenta 25 (2004) 253–258. [52] G.B. Fields, Using fluorogenic peptide substrates to assay matrix
[33] Y. Masui, T. Takemoto, S. Sakakibara, H. Hori, Y. Nagai, Synthetic metalloproteinases, Methods Mol. Biol. 151 (2001) 495–518.
substrates for vertebrate collagenase, Biochem. Med. 17 (1977) 215– [53] B.P. Holskin, M. Bukhtiyarova, B.M. Dunn, P. Baur, J. de Chastonay,
221. M.W. Pennington, A continuous fluorescence-based assay of human
[34] R.D. Gray, H.H. Saneii, Characterization of vertebrate collagenase cytomegalovirus protease using a peptide substrate, Anal. Biochem.
activity by high-performance liquid chromatography using a synthetic 226 (1995) 148–155.
substrate, Anal. Biochem. 120 (1982) 339–346. [54] S. Grahn, D. Ullmann, H.-D. Jakubke, Design and synthesis of fluo-
[35] H.R. Williams, T.Y. Lin, Human polymorphonuclear leukocyte colla- rogenic trypsin peptide substrates based on resonance energy transfer,
genase and gelatinase. Comparison of certain enzymatic properties, Anal. Biochem. 265 (1998) 225–231.
Int. J. Biochem. 16 (1984) 1321–1329. [55] M. Itoh, M. Osaki, T. Chiba, K. Masuda, T. Akizawa, M. Yoshioka,
[36] H. Weingarten, R. Martin, J. Feder, Synthetic substrates of vertebrate et al., Flow injection analysis for measurement of activity of matrix
collagenase, Biochemistry 24 (1985) 6730–6734. metalloproteinase-7 (MMP-7), J. Pharm. Biomed. Anal. 15 (1997)
[37] H. Weingarten, J. Feder, Spectrophotometric assay for vertebrate 1417–1426.
collagenase, Anal. Biochem. 147 (1985) 437–440. [56] C.G. Fields, C.M. Lovdahl, A.J. Miles, V.L. Mathias-Hagen,
[38] C.G. Knight, Fluorimetric assays of proteolytic enzymes, Methods G.B. Fields, Solid-phase synthesis and stability of triple-helical pep-
Enzymol. 248 (1995) 18–34. tides incorporating native collagen sequences, Biopolymers 33 (1993)
[39] M.S. Stack, R.D. Gray, Comparison of vertebrate collagenase and 1695–1707.
gelatinase using a new fluorogenic substrate peptide, J. Biol. Chem. [57] C.G. Fields, D.J. Mickelson, S.L. Drake, J.B. McCarthy, G.B. Fields,
264 (1989) 4277–4281. Melanoma cell adhesion and spreading activities of a synthetic 124-
[40] S. Netzel-Arnett, S.K. Mallya, H. Nagase, H. Birkedal-Hansen, residue triple-helical ‘mini-collagen’, J. Biol. Chem. 268 (1993)
H.E. Van Wart, Continuously recording fluorescent assays optimized 14153–14160.
for five human matrix metalloproteinases, Anal. Biochem. 195 (1991) [58] Y.-C. Yu, P. Berndt, M. Tirrell, G.B. Fields, Self-assembling
86–92. amphiphiles for construction of protein molecular architecture, J. Am.
[41] C.G. Knight, F. Willenbrock, G. Murphy, A novel coumarin-labelled Chem. Soc. 118 (1996) 12515–12520.
peptide for sensitive continuous assays of matrix metalloproteinases, [59] Y.-C. Yu, M. Tirrell, G.B. Fields, Minimal lipidation stabilizes
FEBS Lett. 296 (1992) 263–266. protein-like molecular architecture, J. Am. Chem. Soc. 120 (1998)
[42] D.M. Bickett, M.D. Green, J. Berman, M. Dezube, A.S. Howe, 9979–9987.
P.J. Brown, et al., A high throughput fluorogenic substrate for intersti- [60] Y.-C. Yu, V. Roontga, V.A. Daragan, K.H. Mayo, M. Tirrell,
tial collagenase (MMP-1) and gelatinase (MMP-9), Anal. Biochem. G.B. Fields, Structure and dynamics of peptide amphiphiles incorpo-
212 (1993) 58–64. rating triple-helical protein like molecular architecture, Biochemistry
[43] U. Neumann, H. Kubota, K. Frei, V. Ganu, D. Leppert, Characteriza- 38 (1999) 1659–1668.
tion of Mca-Lys-Pro-Leu-Gly-Leu-Dpa-Ala-Arg-NH2, a fluorogenic [61] J.L. Lauer-Fields, H. Nagase, G.B. Fields, Use of Edman degradation
substrate with increased specificity constants for collagenases and sequence analysis and matrix-assisted laser desorption/ionization
tumor necrosis factor converting enzyme, Anal. Biochem. 328 (2004) mass spectrometry in designing substrates for matrix metalloprotein-
166–173. ases, J. Chromatogr. A 890 (2000) 117–125.
[44] E.D. Matayoshi, G.T. Wang, G.A. Krafft, J. Erickson, Novel fluoro- [62] J.L. Lauer-Fields, K.A. Tuzinski, K. Shimokawa, H. Nagase,
genic substrates for assaying retroviral proteases by resonance energy G.B. Fields, Hydrolysis of triple-helical collagen peptide models by
transfer, Science 247 (1990) 954–958. matrix metalloproteinases, J. Biol. Chem. 275 (2000) 13282–13290.
[45] J.W. Drijfhout, J. Nagel, B. Beekman, J.M. Te Koppele, W. Bloem- [63] J.L. Lauer-Fields, T. Broder, T. Sritharan, L. Chung, H. Nagase,
hoff, Solid phase synthesis of peptides containing the fluorescence G.B. Fields, Kinetic analysis of matrix metalloproteinase activity
energy transfer Dabcyl–Edans couple, in: P.T.P. Kaumaya, using fluorogenic triple-helical substrates, Biochemistry 40 (2001)
R.S. Hodges (Eds.), “Peptides: Chemistry, Structure and Biology”, 5795–5803.
1996, pp. 129–131 pp. [64] J. Ottl, R. Battistuta, M. Pieper, H. Tschesche, W. Bode, K. Kühn,
[46] B. Beekman, J.W. Drijfhout, W. Bloemhoff, H.K. Ronday, P.P. Tak, et al., Design and synthesis of heterotrimeric collagen peptides with a
J.M. Te Koppele, Convenient fluorometric assay for matrix metallo- built-in cystine-knot. Models for collagen catabolism by matrix-
proteinase activity and its application in biological media, FEBS Lett. metalloproteases, FEBS Lett. 398 (1996) 31–36.
390 (1996) 221–225. [65] J. Ottl, L. Moroder, Disulfide-bridged heterotrimeric collagen pep-
[47] B. Beekman, B. van El, J.W. Drijfhout, H.K. Ronday, J.M. Te Kop- tides containing the collagenase cleavage site of collagen type I.
pele, Highly increased levels of active stromelysin in rheumatoid Synthesis and conformational properties, J. Am. Chem. Soc. 121
synovial fluid determined by a selective fluorogenic assay, FEBS Lett. (1999) 653–661.
418 (1997) 305–309. [66] J.C.D. Müller, J. Ottl, L. Moroder, Heterotrimeric collagen peptides as
[48] K.F. Geoghegan, M.J. Emery, W.H. Martin, A.S. McColl, fluorogenic collagenase substrates: synthesis, conformational proper-
G.O. Daumy, Site-directed double fluorescent tagging of human renin ties, and enzymatic digestion, Biochemistry 39 (2000) 5111–5116.
and collagenase (MMP-1) substrate peptides using the periodate oxi- [67] J.L. Lauer-Fields, D. Juska, G.B. Fields, Matrix metalloproteinases
dation of N-terminal serine. An apparently general strategy for provi- and collagen catabolism, Biopolymers 66 (2002) 19–32.
sion of energy-transfer substrates for proteases, Bioconjug. Chem. 4 [68] J.L. Lauer-Fields, G.B. Fields, Triple-helical peptide analysis of col-
(1993) 537–544. lagenolytic protease activity, Biol. Chem. 383 (2002) 1095–1105.
272 C. Lombard et al. / Biochimie 87 (2005) 265–272

[69] J.L. Lauer-Fields, T. Sritharan, M.S. Stack, H. Nagase, G.B. Fields, [73] S. Marini, G.F. Fasciglione, G. de Sanctis, S. D’Alessio, V. Politi,
Selective hydrolysis of triple-helical substrates by matrix metallopro- M. Coletta, Cleavage of bovine collagen I by neutrophil collagenase
teinase-2 and -9, J. Biol. Chem. 278 (2003) 18140–18145. MMP-8. Effect of pH on the catalytic properties as compared to
synthetic substrates, J. Biol. Chem. 275 (2000) 18657–18663.
[70] J.L. Lauer-Fields, P. Kele, G. Sui, H. Nagase, R.M. Leblanc, [74] B. Beekman, J.W. Drijfhout, H.K. Ronday, J.M. Te Koppele, Fluoro-
G.B. Fields, Analysis of matrix metalloproteinase triple-helical pepti- genic MMP activity assay for plasma including MMPs complexed to
dase activity with substrates incorporating fluorogenic L- or D- amino alpha 2-macroglobulin, Ann. N. Y. Acad. Sci. 878 (1999) 150–158.
acids, Anal. Biochem. 321 (2003) 105–115. [75] A.R. Quesada, M.M. Barbacid, E. Mira, P. Fernandez-Resa, G. Mar-
quez, M. Aracil, Evaluation of fluorometric and zymographic methods
[71] D. Minond, J.L. Lauer-Fields, H. Nagase, G.B. Fields, Matrix metal- as activity assays for stromelysins and gelatinases, Clin. Exp.
loproteinase triple-helical peptidase activities are differentially regu- Metastasis 15 (1997) 26–32.
lated by substrate stability, Biochemistry 43 (2004) 11474–11481. [76] F.H. Rasmussen, N. Yeung, L. Kiefer, G. Murphy, C. Lopez-Otin,
M.P. Vitek, et al., Use of a multiple-enzyme/multiple-reagent assay
[72] L. Chung, D. Dinakarpandian, N. Yoshida, J.L. Lauer-Fields, system to quantify activity levels in samples containing mixtures of
G.B. Fields, R. Visse, et al., Collagenase unwinds triple-helical col- matrix metalloproteinases, Biochemistry 43 (2004) 2987–2995.
lagen prior to peptide bond hydrolysis, EMBO J. 23 (2004) 3020– [77] C. Lopez-Otin, C.M. Overall, Protease degradomics: a new challenge
3030. for proteomics, Nat. Rev. Mol. Cell Biol. 3 (2002) 509–519.

View publication stats

Você também pode gostar